1
|
Que X, Zhang T, Liu X, Yin Y, Xia X, Gong P, Song W, Qin Q, Xu ZQD, Tang Y. The role of TREM2 in myelin sheath dynamics: A comprehensive perspective from physiology to pathology. Prog Neurobiol 2025; 247:102732. [PMID: 40021075 DOI: 10.1016/j.pneurobio.2025.102732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/31/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Demyelinating disorders, characterizing by the loss of myelin integrity, present significant challenges due to their impact on neurological function and lack of effective treatments. Understanding the mechanisms underlying myelin damage is crucial for developing therapeutic strategies. Triggering receptor expressed on myeloid cells 2 (TREM2), a pivotal immune receptor predominantly found on microglial cells, plays essential roles in phagocytosis and lipid metabolism, vital processes in neuroinflammation and immune regulation. Emerging evidence indicates a close relationship between TREM2 and various aspects of myelin sheath dynamics, including maintenance, response to damage, and regeneration. This review provides a comprehensive discussion of TREM2's influence on myelin physiology and pathology, highlighting its therapeutic potential and putative mechanisms in the progression of demyelinating disorders.
Collapse
Affiliation(s)
- Xinwei Que
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China; Departments of Neurobiology and Pathology, Capital Medical University, Beijing 100069, China
| | - Tongtong Zhang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | - Xueyu Liu
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | - Yunsi Yin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | - Xinyi Xia
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | - Ping Gong
- Departments of Neurobiology and Pathology, Capital Medical University, Beijing 100069, China
| | - Weiyi Song
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China; Departments of Neurobiology and Pathology, Capital Medical University, Beijing 100069, China
| | - Qi Qin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China.
| | - Zhi-Qing David Xu
- Departments of Neurobiology and Pathology, Capital Medical University, Beijing 100069, China.
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China.
| |
Collapse
|
2
|
Liu P, Zhang K, Tong C, Liu T, Zheng J. Progesterone alleviates esketamine-induced hypomyelination via PI3K/Akt signaling pathway in the developing rat brain. Biotechnol Genet Eng Rev 2024; 40:1202-1217. [PMID: 36946765 DOI: 10.1080/02648725.2023.2193058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
The neurodevelopmental toxicity of anesthetics has been confirmed repeatedly, and esketamine is now widely used in pediatric surgeries. Oligodendrocyte precursor cells (OPCs) evolved into mature oligodendrocytes (OLs) and formed myeline sheath during the early brain development. In this study, we investigated whether esketamine exposure interrupted development of OPCs and induced hypomyelination in rats. Further we explored the roles of PI3K/Akt phosphorylation in OPCs development and myelination. Sprague Dawley rats with different ages (postnatal day (P) 1, 3, 7 and 12) were exposed to 40mg/kg esketamine. Progesterone treatment was given (16 mg/kg per day for 3 days) 24 h after esketamine exposure via the intraperitoneal route. Corpus callosum tissues were collected at P8 or P14 for western blot and immunofluorescence analyses. Esketamine exposure at P7 and P12 significantly reduced myelin basic protein (MBP) expression and CC1+ OLs number in corpus callosum. Esketamine exposure at P7 not only aggravated the mature OLs apoptosis, also decreased the OPCs proliferation and differentiation, which was related with dephosphorylation of PI3K/Akt. Progesterone was able to promote OPCs differentiation and ameliorate esketamine-induced hypomyelination by enhancing PI3K/Akt phosphorylation. Stage-dependent abnormality of OPCs/OLs after esketamine leads to the esketamine-induced hypomyelination. Esketamine interrupted OPCs evolution via PI3K/Akt signaling pathway, which can be ameliorated by progesterone.
Collapse
Affiliation(s)
- Peiwen Liu
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kan Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaoyang Tong
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Liu
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jijian Zheng
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Matteoli M. The role of microglial TREM2 in development: A path toward neurodegeneration? Glia 2024; 72:1544-1554. [PMID: 38837837 DOI: 10.1002/glia.24574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
The nervous and the immune systems undergo a continuous cross talk, starting from early development and continuing throughout adulthood and aging. Defects in this cross talk contribute to neurodevelopmental and neurodegenerative diseases. Microglia are the resident immune cells in the brain that are primarily involved in this bidirectional communication. Among the microglial genes, trem2 is a key player, controlling the functional state of microglia and being at the forefront of many processes that require interaction between microglia and other brain components, such as neurons and oligodendrocytes. The present review focuses on the early developmental window, describing the early brain processes in which TREM2 is primarily involved, including the modulation of synapse formation and elimination, the control of neuronal bioenergetic states as well as the contribution to myelination processes and neuronal circuit formation. By causing imbalances during these early maturation phases, dysfunctional TREM2 may have a striking impact on the adult brain, making it a more sensitive target for insults occurring during adulthood and aging.
Collapse
Affiliation(s)
- Michela Matteoli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Neuro Center, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
4
|
de Almeida V, Seabra G, Reis-de-Oliveira G, Zuccoli GS, Rumin P, Fioramonte M, Smith BJ, Zuardi AW, Hallak JEC, Campos AC, Crippa JA, Martins-de-Souza D. Cannabinoids modulate proliferation, differentiation, and migration signaling pathways in oligodendrocytes. Eur Arch Psychiatry Clin Neurosci 2022; 272:1311-1323. [PMID: 35622101 DOI: 10.1007/s00406-022-01425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/02/2022] [Indexed: 11/03/2022]
Abstract
Cannabinoid signaling, mainly via CB1 and CB2 receptors, plays an essential role in oligodendrocyte health and functions. However, the specific molecular signals associated with the activation or blockade of CB1 and CB2 receptors in this glial cell have yet to be elucidated. Mass spectrometry-based shotgun proteomics and in silico biology tools were used to determine which signaling pathways and molecular mechanisms are triggered in a human oligodendrocytic cell line (MO3.13) by several pharmacological stimuli: the phytocannabinoid cannabidiol (CBD); CB1 and CB2 agonists ACEA, HU308, and WIN55, 212-2; CB1 and CB2 antagonists AM251 and AM630; and endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG). The modulation of cannabinoid signaling in MO3.13 was found to affect pathways linked to cell proliferation, migration, and differentiation of oligodendrocyte progenitor cells. Additionally, we found that carbohydrate and lipid metabolism, as well as mitochondrial function, were modulated by these compounds. Comparing the proteome changes and upstream regulators among treatments, the highest overlap was between the CB1 and CB2 antagonists, followed by overlaps between AEA and 2-AG. Our study opens new windows of opportunities, suggesting that cannabinoid signaling in oligodendrocytes might be relevant in the context of demyelinating and neurodegenerative diseases. Proteomics data are available at ProteomeXchange (PXD031923).
Collapse
Affiliation(s)
- Valéria de Almeida
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil.
| | - Gabriela Seabra
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Guilherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Priscila Rumin
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Mariana Fioramonte
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Bradley J Smith
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Antonio W Zuardi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Jaime E C Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Alline C Campos
- National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - José A Crippa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil. .,Instituto Nacional de Biomarcadores Em Neuropsiquiatria (INBION) Conselho Nacional de Desenvolvimento Científico E Tecnológico, São Paulo, Brazil. .,Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, Brazil. .,D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.
| |
Collapse
|
5
|
Paes de Faria J, Vale-Silva RS, Fässler R, Werner HB, Relvas JB. Pinch2 regulates myelination in the mouse central nervous system. Development 2022; 149:275524. [DOI: 10.1242/dev.200597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/16/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The extensive morphological changes of oligodendrocytes during axon ensheathment and myelination involve assembly of the Ilk-Parvin-Pinch (IPP) heterotrimeric complex of proteins to relay essential mechanical and biochemical signals between integrins and the actin cytoskeleton. Binding of Pinch1 and Pinch2 isoforms to Ilk is mutually exclusive and allows the formation of distinct IPP complexes with specific signaling properties. Using tissue-specific conditional gene ablation in mice, we reveal an essential role for Pinch2 during central nervous system myelination. Unlike Pinch1 gene ablation, loss of Pinch2 in oligodendrocytes results in hypermyelination and in the formation of pathological myelin outfoldings in white matter regions. These structural changes concur with inhibition of Rho GTPase RhoA and Cdc42 activities and phenocopy aspects of myelin pathology observed in corresponding mouse mutants. We propose a dual role for Pinch2 in preventing an excess of myelin wraps through RhoA-dependent control of membrane growth and in fostering myelin stability via Cdc42-dependent organization of cytoskeletal septins. Together, these findings indicate that IPP complexes containing Pinch2 act as a crucial cell-autonomous molecular hub ensuring synchronous control of key signaling networks during developmental myelination.
Collapse
Affiliation(s)
- Joana Paes de Faria
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto 1 , 4200-135 Porto , Portugal
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto 2 , 4200-135 Porto , Portugal
| | - Raquel S. Vale-Silva
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto 1 , 4200-135 Porto , Portugal
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto 2 , 4200-135 Porto , Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto 3 , 4050-313 Porto , Portugal
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry 4 , 82152 Martinsried , Germany
| | - Hauke B. Werner
- Max Planck Institute of Experimental Medicine 5 Department of Neurogenetics , , D-37075 Gottingen , Germany
| | - João B. Relvas
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto 1 , 4200-135 Porto , Portugal
- Department of Neurobiology and Neurological Disease, Glial Cell Biology Laboratory, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto 2 , 4200-135 Porto , Portugal
- Faculty of Medicine, Universidade do Porto 6 Department of Biomedicine , , 4200-319 Porto , Portugal
| |
Collapse
|
6
|
Ortega MA, Chaowen C, Fraile-Martinez O, García-Montero C, Saez MA, Cruza I, Pereda-Cerquella C, Alvarez-Mon MA, Guijarro LG, Fatych Y, Menor-Salván C, Alvarez-Mon M, De Leon-Luis J, Buján J, Garcia-Honduvilla N, Bravo C, Asúnsolo-del-Barco A. Chronic Venous Disease in Pregnant Women Causes an Increase in ILK in the Placental Villi Associated with a Decrease in E-Cadherin. J Pers Med 2022; 12:jpm12020277. [PMID: 35207765 PMCID: PMC8875350 DOI: 10.3390/jpm12020277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
Chronic venous disease (CVD) is a multifactorial vascular disorder frequently manifested in lower limbs in the form of varicose veins (VVs). Women are a vulnerable population for suffering from CVD, especially during pregnancy, when a plethora of changes occur in their cardiovascular system. Previous studies have indicated a worrisome association between CVD in pregnancy with the placental structure and function. Findings include an altered cellular behavior and extracellular matrix (ECM) composition. Integrin-linked kinase (ILK) is a critical molecule involved in multiple physiological and pathological conditions, and together with cadherins, is essential to mediate cell to ECM and cell to cell interplay, respectively. Thus, the aim of this study was to evaluate the implication of ILK and a set of cadherins (e-cadherin, cadherin-6 and cadherin-17) in placentas of women with CVD in order to unravel the possible pathophysiological role of these components. Gene expression (RT-qPCR) and protein expression (immunohistochemistry) studies were performed. Our results show a significant increase in the gene and protein expression of ILK, cadherin-6 and cadherin-17 and a decrease of e-cadherin in the placenta of women with CVD. Overall, this work shows that an abnormal expression of ILK, e-cadherin, cadherin-6 and cadherin-17 may be implicated in the pathological changes occurring in the placental tissue. Further studies should be conducted to determine the possible associations of these changes with maternal and fetal well-being.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28801 Alcalá de Henares, Spain
- Correspondence: (M.A.O.); (C.B.); Tel.: +34-91-885-45-40 (M.A.O.); Fax: +34-91-885-48-85 (M.A.O.)
| | - Chen Chaowen
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Cielo García-Montero
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Miguel A. Saez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Pathological Anatomy Service, Central University Hospital of Defence-UAH, 28001 Madrid, Spain
| | - Iris Cruza
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
| | - Claude Pereda-Cerquella
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
| | - Miguel Angel Alvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain; (Y.F.); (C.M.-S.)
| | - Yuliia Fatych
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain; (Y.F.); (C.M.-S.)
| | - César Menor-Salván
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain; (Y.F.); (C.M.-S.)
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Immune System Diseases-Rheumatology and Oncology Service, University Hospital Príncipe de Asturias, CIBEREHD, 28801 Alcalá de Henares, Spain
| | - Juan De Leon-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Natalio Garcia-Honduvilla
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (M.A.S.); (I.C.); (C.P.-C.); (M.A.A.-M.); (M.A.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
- Correspondence: (M.A.O.); (C.B.); Tel.: +34-91-885-45-40 (M.A.O.); Fax: +34-91-885-48-85 (M.A.O.)
| | - Angel Asúnsolo-del-Barco
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), 28001 Madrid, Spain; (L.G.G.); (A.A.-d.-B.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain;
| |
Collapse
|
7
|
Wang Y, Guo F. Group I PAKs in myelin formation and repair of the central nervous system: what, when, and how. Biol Rev Camb Philos Soc 2021; 97:615-639. [PMID: 34811887 DOI: 10.1111/brv.12815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/20/2021] [Accepted: 11/04/2021] [Indexed: 11/30/2022]
Abstract
p21-activated kinases (PAKs) are a family of cell division control protein 42/ras-related C3 botulinum toxin substrate 1 (Cdc42/Rac1)-activated serine/threonine kinases. Group I PAKs (PAK1-3) have distinct activation mechanisms from group II PAKs (PAK4-6) and are the focus of this review. In transformed cancer cells, PAKs regulate a variety of cellular processes and molecular pathways which are also important for myelin formation and repair in the central nervous system (CNS). De novo mutations in group I PAKs are frequently seen in children with neurodevelopmental defects and white matter anomalies. Group I PAKs regulate virtually every aspect of neuronal development and function. Yet their functions in CNS myelination and remyelination remain incompletely defined. Herein, we highlight the current understanding of PAKs in regulating cellular and molecular pathways and discuss the status of PAK-regulated pathways in oligodendrocyte development. We point out outstanding questions and future directions in the research field of group I PAKs and oligodendrocyte development.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurology, Shriners Hospitals for Children/School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), University of California, Davis, 2425 Stockton Blvd, Sacramento, CA, 95817, U.S.A
| | - Fuzheng Guo
- Department of Neurology, Shriners Hospitals for Children/School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), University of California, Davis, 2425 Stockton Blvd, Sacramento, CA, 95817, U.S.A
| |
Collapse
|
8
|
Jadhav VS, Lin PBC, Pennington T, Di Prisco GV, Jannu AJ, Xu G, Moutinho M, Zhang J, Atwood BK, Puntambekar SS, Bissel SJ, Oblak AL, Landreth GE, Lamb BT. Trem2 Y38C mutation and loss of Trem2 impairs neuronal synapses in adult mice. Mol Neurodegener 2020; 15:62. [PMID: 33115519 PMCID: PMC7594478 DOI: 10.1186/s13024-020-00409-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Triggering receptor expressed on myeloid cells 2 (TREM2) is expressed in the brain exclusively on microglia and genetic variants are linked to neurodegenerative diseases including Alzheimer's disease (AD), frontotemporal dementia (FTD) and Nasu Hakola Disease (NHD). The Trem2 variant R47H, confers substantially elevated risk of developing late onset Alzheimer's disease, while NHD-linked Trem2 variants like Y38C, are associated with development of early onset dementia with white matter pathology. However, it is not known how these Trem2 species, predisposes individuals to presenile dementia. METHODS To investigate if Trem2 Y38C or loss of Trem2 alters neuronal function we generated a novel mouse model to introduce the NHD Trem2 Y38C variant in murine Trem2 using CRISPR/Cas9 technology. Trem2Y38C/Y38C and Trem2-/- mice were assessed for Trem2 expression, differentially expressed genes, synaptic protein levels and synaptic plasticity using biochemical, electrophysiological and transcriptomic approaches. RESULTS While mice harboring the Trem2 Y38C exhibited normal expression levels of TREM2, the pathological outcomes phenocopied Trem2-/- mice at 6 months. Transcriptomic analysis revealed altered expression of neuronal and oligodendrocytes/myelin genes. We observed regional decreases in synaptic protein levels, with the most affected synapses in the hippocampus. These alterations were associated with reduced synaptic plasticity. CONCLUSION Our findings provide in vivo evidence that Trem2 Y38C disrupts normal TREM2 functions. Trem2Y38C/Y38C and Trem2-/- mice demonstrated altered gene expression, changes in microglia morphology, loss of synaptic proteins and reduced hippocampal synaptic plasticity at 6 months in absence of any pathological triggers like amyloid. This suggests TREM2 impacts neuronal functions providing molecular insights on the predisposition of individuals with TREM2 variants resulting in presenile dementia.
Collapse
Affiliation(s)
- Vaishnavi S Jadhav
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Peter B C Lin
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Taylor Pennington
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Gonzalo Viana Di Prisco
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Asha Jacob Jannu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 462020, USA
| | - Guixiang Xu
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Miguel Moutinho
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Anatomy and Cell Biology, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Brady K Atwood
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Shweta S Puntambekar
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Stephanie J Bissel
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Adrian L Oblak
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Radiology & Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Gary E Landreth
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
- Department of Anatomy and Cell Biology, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA
| | - Bruce T Lamb
- Paul and Carole Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
9
|
Roles of Progesterone, Testosterone and Their Nuclear Receptors in Central Nervous System Myelination and Remyelination. Int J Mol Sci 2020; 21:ijms21093163. [PMID: 32365806 PMCID: PMC7246940 DOI: 10.3390/ijms21093163] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
Progesterone and testosterone, beyond their roles as sex hormones, are neuroactive steroids, playing crucial regulatory functions within the nervous system. Among these, neuroprotection and myelin regeneration are important ones. The present review aims to discuss the stimulatory effects of progesterone and testosterone on the process of myelination and remyelination. These effects have been demonstrated in vitro (i.e., organotypic cultures) and in vivo (cuprizone- or lysolecithin-induced demyelination and experimental autoimmune encephalomyelitis (EAE)). Both steroids stimulate myelin formation and regeneration by acting through their respective intracellular receptors: progesterone receptors (PR) and androgen receptors (AR). Activation of these receptors results in multiple events involving direct transcription and translation, regulating general homeostasis, cell proliferation, differentiation, growth and myelination. It also ameliorates immune response as seen in the EAE model, resulting in a significant decrease in inflammation leading to a fast recovery. Although natural progesterone and testosterone have a therapeutic potential, their synthetic derivatives—the 19-norprogesterone (nestorone) and 7α-methyl-nortestosterone (MENT), already used as hormonal contraception or in postmenopausal hormone replacement therapies, may offer enhanced benefits for myelin repair. We summarize here a recent advancement in the field of myelin biology, to treat demyelinating disorders using the natural as well as synthetic analogs of progesterone and testosterone.
Collapse
|
10
|
Jiang C, Zeng X, Xue B, Campbell D, Wang Y, Sun H, Xu Y, Wen X. Screening of pure synthetic coating substrates for induced pluripotent stem cells and iPSC-derived neuroepithelial progenitors with short peptide based integrin array. Exp Cell Res 2019; 380:90-99. [PMID: 30981669 DOI: 10.1016/j.yexcr.2019.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 01/27/2023]
Abstract
Simple and pure synthetic coating substrates are needed to overcome the disadvantages of traditional coating products like animal derived Matrigel in stem cell research. Since integrins are of great importance in cell adhesion and cell-ECM communication, in this study, a commercially available integrin array established by synthetic integrin binding peptides is used to screen coating substrates for iPSCs and NEPs. The results showed that binding peptides of integrin α5β1, αVβ1, αMβ2 and αIIbβ3 supported cell adhesion of iPSCs, while α5β1, αVβ1 and αIIbβ3 binding peptides supported NEPs adhesion. Additionally, integrin α5β1 binding peptide was revealed to support rapid expansion of iPSCs and iPSC-derived NEPs, as well as the process of NEPs generation, with equal efficiency as Matrigel. In this work, we demonstrated that by supporting stem cell growth in an integrin dependent manner, the integrin array and coating system has the potential to develop more precise and efficient systems in neurological disease modeling.
Collapse
Affiliation(s)
- Chenyang Jiang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Xiaomei Zeng
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Bo Xue
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Debbie Campbell
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Yanlin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Huifang Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Xuejun Wen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, 23220, USA; School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China; Shanghai East Hospital, Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
11
|
Cytoskeletal Signal-Regulated Oligodendrocyte Myelination and Remyelination. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:33-42. [DOI: 10.1007/978-981-32-9636-7_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
12
|
Hussain R, Zubair H, Pursell S, Shahab M. Neurodegenerative Diseases: Regenerative Mechanisms and Novel Therapeutic Approaches. Brain Sci 2018; 8:E177. [PMID: 30223579 PMCID: PMC6162719 DOI: 10.3390/brainsci8090177] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/03/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022] Open
Abstract
Regeneration refers to regrowth of tissue in the central nervous system. It includes generation of new neurons, glia, myelin, and synapses, as well as the regaining of essential functions: sensory, motor, emotional and cognitive abilities. Unfortunately, regeneration within the nervous system is very slow compared to other body systems. This relative slowness is attributed to increased vulnerability to irreversible cellular insults and the loss of function due to the very long lifespan of neurons, the stretch of cells and cytoplasm over several dozens of inches throughout the body, insufficiency of the tissue-level waste removal system, and minimal neural cell proliferation/self-renewal capacity. In this context, the current review summarized the most common features of major neurodegenerative disorders; their causes and consequences and proposed novel therapeutic approaches.
Collapse
Affiliation(s)
- Rashad Hussain
- Center for Translational Neuromedicine, University of Rochester, NY 14642, USA.
| | - Hira Zubair
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| | - Sarah Pursell
- Center for Translational Neuromedicine, University of Rochester, NY 14642, USA.
| | - Muhammad Shahab
- Department of Animal Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|